Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Dis Model Mech ; 17(7)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38804708

RESUMEN

The TATA box-binding protein-associated factor 1 (TAF1) is a ubiquitously expressed protein and the largest subunit of the basal transcription factor TFIID, which plays a key role in initiation of RNA polymerase II-dependent transcription. TAF1 missense variants in human males cause X-linked intellectual disability, a neurodevelopmental disorder, and TAF1 is dysregulated in X-linked dystonia-parkinsonism, a neurodegenerative disorder. However, this field has lacked a genetic mouse model of TAF1 disease to explore its mechanism in mammals and treatments. Here, we generated and validated a conditional cre-lox allele and the first ubiquitous Taf1 knockout mouse. We discovered that Taf1 deletion in male mice was embryonically lethal, which may explain why no null variants have been identified in humans. In the brains of Taf1 heterozygous female mice, no differences were found in gross structure, overall expression and protein localisation, suggesting extreme skewed X inactivation towards the non-mutant chromosome. Nevertheless, these female mice exhibited a significant increase in weight, weight with age, and reduced movement, suggesting that a small subset of neurons was negatively impacted by Taf1 loss. Finally, this new mouse model may be a future platform for the development of TAF1 disease therapeutics.


Asunto(s)
Peso Corporal , Heterocigoto , Histona Acetiltransferasas , Ratones Noqueados , Trastornos del Movimiento , Factores Asociados con la Proteína de Unión a TATA , Factor de Transcripción TFIID , Animales , Factores Asociados con la Proteína de Unión a TATA/genética , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/genética , Factor de Transcripción TFIID/metabolismo , Factor de Transcripción TFIID/deficiencia , Femenino , Masculino , Histona Acetiltransferasas/metabolismo , Histona Acetiltransferasas/genética , Trastornos del Movimiento/genética , Trastornos del Movimiento/patología , Embrión de Mamíferos/metabolismo , Ratones , Encéfalo/patología , Encéfalo/metabolismo , Genes Letales , Ratones Endogámicos C57BL
2.
Neurobiol Dis ; 193: 106437, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38367882

RESUMEN

TDP-43 pathology is found in several neurodegenerative disorders, collectively referred to as "TDP-43 proteinopathies". Aggregates of TDP-43 are present in the brains and spinal cords of >97% of amyotrophic lateral sclerosis (ALS), and in brains of ∼50% of frontotemporal dementia (FTD) patients. While mutations in the TDP-43 gene (TARDBP) are usually associated with ALS, many clinical reports have linked these mutations to cognitive impairments and/or FTD, but also to other neurodegenerative disorders including Parkinsonism (PD) or progressive supranuclear palsy (PSP). TDP-43 is a ubiquitously expressed, highly conserved RNA-binding protein that is involved in many cellular processes, mainly RNA metabolism. To investigate systemic pathological mechanisms in TDP-43 proteinopathies, aiming to capture the pleiotropic effects of TDP-43 mutations, we have further characterised a mouse model carrying a point mutation (M323K) within the endogenous Tardbp gene. Homozygous mutant mice developed cognitive and behavioural deficits as early as 3 months of age. This was coupled with significant brain structural abnormalities, mainly in the cortex, hippocampus, and white matter fibres, together with progressive cortical interneuron degeneration and neuroinflammation. At the motor level, progressive phenotypes appeared around 6 months of age. Thus, cognitive phenotypes appeared to be of a developmental origin with a mild associated progressive neurodegeneration, while the motor and neuromuscular phenotypes seemed neurodegenerative, underlined by a progressive loss of upper and lower motor neurons as well as distal denervation. This is accompanied by progressive elevated TDP-43 protein and mRNA levels in cortex and spinal cord of homozygous mutant mice from 3 months of age, together with increased cytoplasmic TDP-43 mislocalisation in cortex, hippocampus, hypothalamus, and spinal cord at 12 months of age. In conclusion, we find that Tardbp M323K homozygous mutant mice model many aspects of human TDP-43 proteinopathies, evidencing a dual role for TDP-43 in brain morphogenesis as well as in the maintenance of the motor system, making them an ideal in vivo model system to study the complex biology of TDP-43.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Proteinopatías TDP-43 , Animales , Preescolar , Humanos , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Encéfalo/metabolismo , Cognición , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Proteinopatías TDP-43/genética , Proteinopatías TDP-43/patología
3.
Neuronal Signal ; 8(1): NS20230020, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38222463

RESUMEN

This scientific commentary refers to 'Inhibition of insulin-degrading enzyme in human neurons promotes amyloid-ß deposition' by Rowland et al. (https://doi.org/10.1042/NS20230016). Insulin-degrading enzyme (IDE) and neprilysin (NEP) have been proposed as two Aß-degrading enzymes supported by human genetics and in vivo data. Rowland et al. provide complementary evidence of a key role for IDE in Aß metabolism in human-induced pluripotent stem cell (iPSC)-derived cortical neurons.

4.
Neurobiol Dis ; 190: 106363, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37996040

RESUMEN

Sporadic Creutzfeldt-Jakob disease (sCJD), the most common human prion disease, is thought to occur when the cellular prion protein (PrPC) spontaneously misfolds and assembles into prion fibrils, culminating in fatal neurodegeneration. In a genome-wide association study of sCJD, we recently identified risk variants in and around the gene STX6, with evidence to suggest a causal increase of STX6 expression in disease-relevant brain regions. STX6 encodes syntaxin-6, a SNARE protein primarily involved in early endosome to trans-Golgi network retrograde transport. Here we developed and characterised a mouse model with genetic depletion of Stx6 and investigated a causal role of Stx6 expression in mouse prion disease through a classical prion transmission study, assessing the impact of homozygous and heterozygous syntaxin-6 knockout on disease incubation periods and prion-related neuropathology. Following inoculation with RML prions, incubation periods in Stx6-/- and Stx6+/- mice differed by 12 days relative to wildtype. Similarly, in Stx6-/- mice, disease incubation periods following inoculation with ME7 prions also differed by 12 days. Histopathological analysis revealed a modest increase in astrogliosis in ME7-inoculated Stx6-/- animals and a variable effect of Stx6 expression on microglia activation, however no differences in neuronal loss, spongiform change or PrP deposition were observed at endpoint. Importantly, Stx6-/- mice are viable and fertile with no gross impairments on a range of neurological, biochemical, histological and skeletal structure tests. Our results provide some support for a pathological role of Stx6 expression in prion disease, which warrants further investigation in the context of prion disease but also other neurodegenerative diseases considering syntaxin-6 appears to have pleiotropic risk effects in progressive supranuclear palsy and Alzheimer's disease.


Asunto(s)
Síndrome de Creutzfeldt-Jakob , Enfermedades por Prión , Priones , Ratones , Humanos , Animales , Síndrome de Creutzfeldt-Jakob/genética , Síndrome de Creutzfeldt-Jakob/patología , Priones/genética , Priones/metabolismo , Estudio de Asociación del Genoma Completo , Ratones Transgénicos , Encéfalo/metabolismo , Enfermedades por Prión/genética , Enfermedades por Prión/patología , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo
5.
Dis Model Mech ; 16(10)2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37772684

RESUMEN

Variants in the ubiquitously expressed DNA/RNA-binding protein FUS cause aggressive juvenile forms of amyotrophic lateral sclerosis (ALS). Most FUS mutation studies have focused on motor neuron degeneration; little is known about wider systemic or developmental effects. We studied pleiotropic phenotypes in a physiological knock-in mouse model carrying the pathogenic FUSDelta14 mutation in homozygosity. RNA sequencing of multiple organs aimed to identify pathways altered by the mutant protein in the systemic transcriptome, including metabolic tissues, given the link between ALS-frontotemporal dementia and altered metabolism. Few genes were commonly altered across all tissues, and most genes and pathways affected were generally tissue specific. Phenotypic assessment of mice revealed systemic metabolic alterations related to the pathway changes identified. Magnetic resonance imaging brain scans and histological characterisation revealed that homozygous FUSDelta14 brains were smaller than heterozygous and wild-type brains and displayed significant morphological alterations, including a thinner cortex, reduced neuronal number and increased gliosis, which correlated with early cognitive impairment and fatal seizures. These findings show that the disease aetiology of FUS variants can include both neurodevelopmental and systemic alterations.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ratones , Animales , Esclerosis Amiotrófica Lateral/patología , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Mutación/genética , Neuronas/metabolismo
6.
Mol Neurodegener ; 18(1): 30, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37143081

RESUMEN

Amyotrophic lateral sclerosis is a complex disorder most of which is 'sporadic' of unknown origin but approximately 10% is familial, arising from single mutations in any of more than 30 genes. Thus, there are more than 30 familial ALS subtypes, with different, often unknown, molecular pathologies leading to a complex constellation of clinical phenotypes. We have mouse models for many genetic forms of the disorder, but these do not, on their own, necessarily show us the key pathological pathways at work in human patients. To date, we have no models for the 90% of ALS that is 'sporadic'. Potential therapies have been developed mainly using a limited set of mouse models, and through lack of alternatives, in the past these have been tested on patients regardless of aetiology. Cancer researchers have undertaken therapy development with similar challenges; they have responded by producing complex mouse models that have transformed understanding of pathological processes, and they have implemented patient stratification in multi-centre trials, leading to the effective translation of basic research findings to the clinic. ALS researchers have successfully adopted this combined approach, and now to increase our understanding of key disease pathologies, and our rate of progress for moving from mouse models to mechanism to ALS therapies we need more, innovative, complex mouse models to address specific questions.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ratones , Animales , Humanos , Esclerosis Amiotrófica Lateral/metabolismo , Modelos Animales de Enfermedad , Mutación , Fenotipo
7.
Neuronal Signal ; 6(3): NS20220065, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36238655

RESUMEN

This themed collection of articles was prompted by a collaboration between Neuronal Signaling and the British Neuroscience Association. The Biochemical Society and Portland Press organised a symposium at the BNA Festival of Neuroscience in 2021, focused on the development and use of experimental models of human neuronal disorders. One aspect dealt with how new technologies are being (or could be) used both as a substitute for, or to complement, research that uses whole animal models. Another aspect discussed factors that need to be considered when appraising the validity of animal models of complex, multifactorial neuronal disorders. Given its relevance to the scope of Neuronal Signaling, the journal's Editorial Board developed a themed collection of content around this symposium entitled Emerging technologies for research models of human neuronal disorders in vivo and in vitro. We were delighted that speakers from the symposium and other experts working in this field agreed to submit reviews for the collection, which offers an invaluable resource both for researchers who are already experts in this field and those who need merely to learn about its scope and potential.

8.
Biomedicines ; 10(5)2022 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-35625841

RESUMEN

There is an increasing interest in the study of the relation between alterations in systemic lipid metabolism and neurodegenerative disorders, in particular in Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD). In ALS these alterations are well described and evident not only with the progression of the disease but also years before diagnosis. Still, there are some discrepancies in findings relating to the causal nature of lipid metabolic alterations, partly due to the great clinical heterogeneity in ALS. ALS presentation is within a disorder spectrum with Frontotemporal Dementia (FTD), and many patients present mixed forms of ALS and FTD, thus increasing the variability. Lipid metabolic and other systemic metabolic alterations have not been well studied in FTD, or in ALS-FTD mixed forms, as has been in pure ALS. With the recent development in lipidomics and the integration with other -omics platforms, there is now emerging data that not only facilitates the identification of biomarkers but also enables understanding of the underlying pathological mechanisms. Here, we reviewed the recent literature to compile lipid metabolic alterations in ALS, FTD, and intermediate mixed forms, with a view to appraising key commonalities or differences within the spectrum.

10.
Commun Biol ; 4(1): 1025, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34471224

RESUMEN

Mutations in the RNA-binding protein (RBP) FUS have been genetically associated with the motoneuron disease amyotrophic lateral sclerosis (ALS). Using both human induced pluripotent stem cells and mouse models, we found that FUS-ALS causative mutations affect the activity of two relevant RBPs with important roles in neuronal RNA metabolism: HuD/ELAVL4 and FMRP. Mechanistically, mutant FUS leads to upregulation of HuD protein levels through competition with FMRP for HuD mRNA 3'UTR binding. In turn, increased HuD levels overly stabilize the transcript levels of its targets, NRN1 and GAP43. As a consequence, mutant FUS motoneurons show increased axon branching and growth upon injury, which could be rescued by dampening NRN1 levels. Since similar phenotypes have been previously described in SOD1 and TDP-43 mutant models, increased axonal growth and branching might represent broad early events in the pathogenesis of ALS.


Asunto(s)
Axones/metabolismo , Proteína 4 Similar a ELAV/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína FUS de Unión a ARN/genética , Animales , Línea Celular , Proteína 4 Similar a ELAV/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Ratones , Neuronas Motoras/metabolismo , Mutación , Proteína FUS de Unión a ARN/metabolismo
11.
Sci Rep ; 11(1): 12251, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112844

RESUMEN

The neuromuscular junction (NMJ) is the peripheral synapse formed between a motor neuron axon terminal and a muscle fibre. NMJs are thought to be the primary site of peripheral pathology in many neuromuscular diseases, but innervation/denervation status is often assessed qualitatively with poor systematic criteria across studies, and separately from 3D morphological structure. Here, we describe the development of 'NMJ-Analyser', to comprehensively screen the morphology of NMJs and their corresponding innervation status automatically. NMJ-Analyser generates 29 biologically relevant features to quantitatively define healthy and aberrant neuromuscular synapses and applies machine learning to diagnose NMJ degeneration. We validated this framework in longitudinal analyses of wildtype mice, as well as in four different neuromuscular disease models: three for amyotrophic lateral sclerosis (ALS) and one for peripheral neuropathy. We showed that structural changes at the NMJ initially occur in the nerve terminal of mutant TDP43 and FUS ALS models. Using a machine learning algorithm, healthy and aberrant neuromuscular synapses are identified with 95% accuracy, with 88% sensitivity and 97% specificity. Our results validate NMJ-Analyser as a robust platform for systematic and structural screening of NMJs, and pave the way for transferrable, and cross-comparison and high-throughput studies in neuromuscular diseases.


Asunto(s)
Enfermedades Neuromusculares/etiología , Enfermedades Neuromusculares/metabolismo , Unión Neuromuscular/metabolismo , Animales , Biomarcadores , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Técnica del Anticuerpo Fluorescente , Aprendizaje Automático , Ratones , Ratones Noqueados , Enfermedades Neuromusculares/diagnóstico , Unión Neuromuscular/patología , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Curva ROC
12.
Mamm Genome ; 32(2): 94-103, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33713180

RESUMEN

The small EDRK-rich factor 2 (SERF2) is a highly conserved protein that modifies amyloid fibre assembly in vitro and promotes protein misfolding. However, the role of SERF2 in regulating age-related proteotoxicity remains largely unexplored due to a lack of in vivo models. Here, we report the generation of Serf2 knockout mice using an ES cell targeting approach, with Serf2 knockout alleles being bred onto different defined genetic backgrounds. We highlight phenotyping data from heterozygous Serf2+/- mice, including unexpected male-specific phenotypes in startle response and pre-pulse inhibition. We report embryonic lethality in Serf2-/- null animals when bred onto a C57BL/6 N background. However, homozygous null animals were viable on a mixed genetic background and, remarkably, developed without obvious abnormalities. The Serf2 knockout mice provide a powerful tool to further investigate the role of SERF2 protein in previously unexplored pathophysiological pathways in the context of a whole organism.


Asunto(s)
Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Péptidos y Proteínas de Señalización Intracelular/genética , Fenotipo , Factores de Edad , Alelos , Empalme Alternativo , Animales , Línea Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Femenino , Regulación de la Expresión Génica , Estudios de Asociación Genética/métodos , Antecedentes Genéticos , Sitios Genéticos , Genotipo , Masculino , Ratones , Ratones Noqueados , Especificidad de Órganos , Microtomografía por Rayos X
13.
iScience ; 24(12): 103463, 2021 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-34988393

RESUMEN

Amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) is a fatal neurodegenerative disorder, and continued innovation is needed for improved understanding and for developing therapeutics. We have created next-generation genomically humanized knockin mouse models, by replacing the mouse genomic region of Sod1, Tardbp (TDP-43), and Fus, with their human orthologs, preserving human protein biochemistry and splicing with exons and introns intact. We establish a new standard of large knockin allele quality control, demonstrating the utility of indirect capture for enrichment of a genomic region of interest followed by Oxford Nanopore sequencing. Extensive analysis shows that homozygous humanized animals only express human protein at endogenous levels. Characterization of humanized FUS animals showed that they are phenotypically normal throughout their lifespan. These humanized strains are vital for preclinical assessment of interventions and serve as templates for the addition of coding or non-coding human ALS/FTD mutations to dissect disease pathomechanisms, in a physiological context.

14.
Methods ; 191: 15-22, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32721467

RESUMEN

Aberrant microsatellite repeat-expansions at specific loci within the human genome cause several distinct, heritable, and predominantly neurological, disorders. Creating models for these diseases poses a challenge, due to the instability of such repeats in bacterial vectors, especially with large repeat expansions. Designing constructs for more precise genome engineering projects, such as engineering knock-in mice, proves a greater challenge still, since these unstable repeats require numerous cloning steps in order to introduce homology arms or selection cassettes. Here, we report our efforts to clone a large hexanucleotide repeat in the C9orf72 gene, originating from within a BAC construct, derived from a C9orf72-ALS patient. We provide detailed methods for efficient repeat sizing and growth conditions in bacteria to facilitate repeat retention during growth and sub-culturing. We report that sub-cloning into a linear vector dramatically improves stability, but is dependent on the relative orientation of DNA replication through the repeat, consistent with previous studies. We envisage the findings presented here provide a relatively straightforward route to maintaining large-range microsatellite repeat-expansions, for efficient cloning into vectors.


Asunto(s)
Expansión de las Repeticiones de ADN , Esclerosis Amiotrófica Lateral/genética , Animales , Proteína C9orf72/genética , Clonación Molecular , Marcación de Gen , Humanos , Ratones
16.
Mamm Genome ; 30(7-8): 173-191, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31203387

RESUMEN

Neurodegenerative disease encompasses a wide range of disorders afflicting the central and peripheral nervous systems and is a major unmet biomedical need of our time. There are very limited treatments, and no cures, for most of these diseases, including Alzheimer's Disease, Parkinson's Disease, Huntington Disease, and Motor Neuron Diseases. Mouse and other animal models provide hope by analysing them to understand pathogenic mechanisms, to identify drug targets, and to develop gene therapies and stem cell therapies. However, despite many decades of research, virtually no new treatments have reached the clinic. Increasingly, it is apparent that human heterogeneity within clinically defined neurodegenerative disorders, and between patients with the same genetic mutations, significantly impacts disease presentation and, potentially, therapeutic efficacy. Therefore, stratifying patients according to genetics, lifestyle, disease presentation, ethnicity, and other parameters may hold the key to bringing effective therapies from the bench to the clinic. Here, we discuss genetic and cellular humanised mouse models, and how they help in defining the genetic and environmental parameters associated with neurodegenerative disease, and so help in developing effective precision medicine strategies for future healthcare.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Neurodegenerativas/terapia , Medicina de Precisión , Animales , Quimera , Humanos , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Fenotipo
17.
Nat Commun ; 10(1): 1796, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30996264

RESUMEN

Metabolic reprogramming is an active regulator of stem cell fate choices, and successful stem cell differentiation in different compartments requires the induction of oxidative phosphorylation. However, the mechanisms that promote mitochondrial respiration during stem cell differentiation are poorly understood. Here we demonstrate that Stat3 promotes muscle stem cell myogenic lineage progression by stimulating mitochondrial respiration in mice. We identify Fam3a, a cytokine-like protein, as a major Stat3 downstream effector in muscle stem cells. We demonstrate that Fam3a is required for muscle stem cell commitment and skeletal muscle development. We show that myogenic cells secrete Fam3a, and exposure of Stat3-ablated muscle stem cells to recombinant Fam3a in vitro and in vivo rescues their defects in mitochondrial respiration and myogenic commitment. Together, these findings indicate that Fam3a is a Stat3-regulated secreted factor that promotes muscle stem cell oxidative metabolism and differentiation, and suggests that Fam3a is a potential tool to modulate cell fate choices.


Asunto(s)
Diferenciación Celular , Citocinas/fisiología , Desarrollo de Músculos/fisiología , Mioblastos/fisiología , Factor de Transcripción STAT3/fisiología , Células Madre/fisiología , Animales , Animales Recién Nacidos , Linaje de la Célula/fisiología , Células Cultivadas , Embrión de Mamíferos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Estriado/citología , Músculo Estriado/crecimiento & desarrollo , Fosforilación Oxidativa , Transducción de Señal/fisiología
18.
Nat Commun ; 10(1): 1845, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015419

RESUMEN

To better understand human health and disease, researchers create a wide variety of mouse models that carry human DNA. With recent advances in genome engineering, the targeted replacement of mouse genomic regions with orthologous human sequences has become increasingly viable, ranging from finely tuned humanisation of individual nucleotides and amino acids to the incorporation of many megabases of human DNA. Here, we examine emerging technologies for targeted genomic humanisation, we review the spectrum of existing genomically humanised mouse models and the insights such models have provided, and consider the lessons learned for designing such models in the future.


Asunto(s)
Edición Génica/métodos , Genoma Humano/genética , Ratones/genética , Modelos Animales , Animales , Cromosomas Humanos/genética , Edición Génica/tendencias , Técnicas de Transferencia de Gen/tendencias , Humanos , Ratones Transgénicos , Proyectos de Investigación
20.
Dev Biol ; 441(1): 127-131, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29964026

RESUMEN

In mouse, retinoic acid (RA) is required for the early phase of body axis extension controlled by a population of neuromesodermal progenitors (NMPs) in the trunk called expanding-NMPs, but not for the later phase of body axis extension controlled by a population of NMPs in the tail called depleting-NMPs. Recent observations suggest that zebrafish utilize depleting-NMPs but not expanding-NMPs for body axis extension. In zebrafish, a role for RA in body axis extension was not supported by previous studies on aldh1a2 (raldh2) mutants lacking RA synthesis. Here, by treating zebrafish embryos with an RA synthesis inhibitor, we also found that body axis extension and somitogenesis was not perturbed, although loss of pectoral fin and cardiac edema were observed consistent with previous studies. The conclusion that zebrafish diverges from mouse in not requiring RA for body axis extension is consistent with zebrafish lacking early expanding-NMPs to generate the trunk. We suggest that RA control of body axis extension was added to higher vertebrates during evolution of expanding-NMPs.


Asunto(s)
Embrión de Mamíferos/embriología , Embrión no Mamífero/embriología , Mesodermo/embriología , Células-Madre Neurales/metabolismo , Tretinoina/metabolismo , Pez Cebra/embriología , Animales , Embrión de Mamíferos/citología , Embrión no Mamífero/citología , Mesodermo/citología , Ratones , Células-Madre Neurales/citología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA